Myelofibrosis (MF), including primary MF, postpolycythemia vera MF, and postessential thrombocythemia MF, is a clonal stem cell disorder characterized by BM fibrosis, extramedullary hematopoiesis, and a variable propensity to transform into acute leukemia. Allogeneic stem cell transplantation is the only known cure for MF, but its applicability is limited by the advanced age of most patients and by comorbid conditions. In the past decade, there has been an explosion of information on the molecular-genetic features associated with these diseases, fueled recently by the discovery of the JAK2V617F mutation. The development of JAK inhibitors has represented a significant therapeutic advance for these diseases; however, their use in MF has not yet been associated with eradication or a significant suppression of the malignant clone. In this era, much remains to be understood about MF, but it is likely that the identification of key pathogenetic drivers of the disease, coupled with the availability of novel molecularly targeted agents, will result in the discovery of new agents that significantly alter the natural history of the disease. This review focuses on recent and ongoing efforts in the development of novel agents in MF that go beyond the field of JAK inhibitors.

The discovery of the JAK2V617F mutation in 2005 generated considerable excitement in the field of the Philadelphia-chromosome-negative (Ph) myeloproliferative neoplasms (MPNs) and raised for the first time the question of whether these diseases may fall into the “one initiating mutation, one target” paradigm that has revolutionized the treatment of chronic myeloid leukemia. In the past few years, we have witnessed the clinical development of JAK-ATP mimetic inhibitors for the treatment of myelofibrosis (MF; used in this review to encompass primary MF, postpolycythemia vera MF, and post essential thrombocythemia MF) and have seen the therapeutic benefits for patients treated with these agents.1,2 

It is clear, however, that there are limitations associated with the use of the currently approved JAK inhibitor ruxolitinib, as well as several other JAK inhibitors that are in clinical development. The clinical benefits to date with these agents have centered largely on effective palliation of symptoms and improvement in splenomegaly, although there is emerging evidence of a potential survival benefit with ruxolitinib.3  Significant areas of unmet need in the therapeutic arena include MF associated with significant cytopenias and/or transfusion dependency, accelerated or blast phase disease, and MF patients who progress on JAK inhibitors or who fail to achieve a response or experience clinical benefit with these agents.

At the same time, the molecular complexity of the Ph MPNs has become increasingly obvious (Figure 1) and these findings, along with the availability of novel agents for clinical investigation, is ushering in a new era in which novel targeted approaches can be pursued. Epigenetic therapies and approaches, immunomodulatory agents, agents that target the dysregulated cytokine milieu, and antifibrosis agents are being actively investigated (Table 1). In addition, there are various non-JAK kinase signal transduction inhibitors under investigation. This review focuses on the potential merits of and the experience thus far with these novel therapeutic approaches and highlights areas of ongoing and future investigation.

Figure 1.

Gene mutations in PMF. Pie chart illustrates the molecular heterogeneity of primary MF (PMF) based on mutations in JAK2, ASXL1, TET2, SRSF2, DNMT3A, MPL, EZH2, CBL, IDH1, and IDH2. Asterisks depict mutations contributing to epigenetic dysregulation in PMF and/or occurring in epigenetic modifiers. Overlapping mutations (co-occurrence of 2 or more mutations in patients with PMF) are not depicted on this chart so the percentages add up to more than 100%. Chart is created from data derived from mutational analysis of all 10 markers in a cohort of 483 patients with PMF published by Vannucchi et al.7 

Figure 1.

Gene mutations in PMF. Pie chart illustrates the molecular heterogeneity of primary MF (PMF) based on mutations in JAK2, ASXL1, TET2, SRSF2, DNMT3A, MPL, EZH2, CBL, IDH1, and IDH2. Asterisks depict mutations contributing to epigenetic dysregulation in PMF and/or occurring in epigenetic modifiers. Overlapping mutations (co-occurrence of 2 or more mutations in patients with PMF) are not depicted on this chart so the percentages add up to more than 100%. Chart is created from data derived from mutational analysis of all 10 markers in a cohort of 483 patients with PMF published by Vannucchi et al.7 

Close modal
Table 1.

Selected single-agent clinical trials in MF

Selected single-agent clinical trials in MF
Selected single-agent clinical trials in MF

*References are cited for reports of early-phase experience. The www.ClinicalTrials.gov listings are provided for ongoing trials.

Epigenetic changes have generally been used to refer to heritable changes in gene expression that occur without a change in coding sequence. Epigenetic changes fall broadly into 2 major categories: changes in DNA methylation and changes in histone modifications. For example, recruitment of histone deacetylases (HDACs) and promoter DNA hypermethylation are 2 pathways of epigenetic silencing that are linked and are implicated in the transcriptional dysregulation underlying a variety of myeloid neoplasms. Unlike gene deletions, which lead to irreversible loss of function, transcriptional dysregulation by histone deacetylation or DNA methylation can potentially be reversed by chromatin-remodeling agents such as HDAC and DNA methyltransferase (DNMT) inhibitors, restoring tumor cells to a more transcriptionally normal state.4 

For many years, there was a paucity of information on the scope of epigenetic changes in MF, and most of the initial studies published focused on analysis of the methylation status of individual genes of interest. Some of these studies provided evidence that epigenetic mechanisms silence genes that inhibit cellular proliferation in MF, such as the SOCS genes, which negatively regulate JAK/STAT signaling,5  or genes such as CXCR4,6  which regulates homing of stem cells to the BM. Perhaps one of the most compelling reasons to evoke epigenetic dysregulation as a central aspect of MF pathogenesis is the fact that, within the past few years, mutations in several genes that encode for proteins with a critical role in modifying the epigenome have been described (Figure 1).7  In addition, both wild-type and mutant JAK2 kinases have been demonstrated to participate in posttranslational modification of specific histone residues.8,9 

Recently, MF has also been shown to have a distinct methylation signature (compared with normal controls and other Ph MPNs) that consists of both aberrantly hypomethylated and hypermethylated loci.10  Pathways that were targeted by hypomethylation included genes affecting cell signaling, hematopoiesis, and immunologic pathways, whereas those that were targeted by hypermethylation included genes involved in inflammatory responses.

Overall, these findings raise the question of the clinical relevance of using chromatin-modifying agents or epigenetic modulators in MF. The potential promise of this approach has been suggested by the preclinical experience with these agents. For example, the use of an HDAC inhibitor in combination with a DNMT inhibitor in vitro led to a reduction in the numbers of MF progenitor CD34+ cells independently of the JAK2 mutational status. This is in contrast to the effects of these agents on normal primitive hematopoietic progenitor (CD34+) cells, where an expansion of these cells has been observed. The reduction in MF CD34+ cells was associated with up-regulation of CXCR4 transcript levels. In a NOD/SCID mouse model, these effects were associated with correction of the abnormal stem cell trafficking associated with this disease, resulting in homing of the cells to the BM, rather than to the spleen.11 

HDAC inhibitors as single agents have been demonstrated in vitro to cause a down-modulation of JAK2V617F protein expression in JAK2 mutant cell lines and primary patient samples, but had no effect on the wild-type JAK2 protein.12  There was no effect on JAK2 mutant and wild-type transcript levels, suggesting that this effect was at the posttranscriptional level. It is likely that these inhibitory effects on the JAK2V617F protein were mediated at least in part through inhibition of HDAC6 and acetylation of HSP90,13  with resultant degradation of the JAK2 mutant protein (Figure 2). Both wild-type and mutant JAK2 proteins are known clients of HSP90, and the mutant protein may be more sensitive to degradation by the ubiquitin proteosome system in the context of disruption of the chaperone function of HSP90.14 

Figure 2.

Putative mechanism of action of HDAC inhibitors in MF. HSP90 is known to associate with HDAC6. JAK2 proteins (both wild-type and mutant proteins) are known clients of HSP90, which stabilizes these proteins. The use of a HDAC inhibitor leads to inhibition of HDAC6, with subsequent acetylation of HSP90 and resultant targeting of JAK2 proteins for proteosomal degradation. This mechanism may contribute at least in part to the down-modulation of the JAK2V617F protein observed with the use of HDAC inhibitors.

Figure 2.

Putative mechanism of action of HDAC inhibitors in MF. HSP90 is known to associate with HDAC6. JAK2 proteins (both wild-type and mutant proteins) are known clients of HSP90, which stabilizes these proteins. The use of a HDAC inhibitor leads to inhibition of HDAC6, with subsequent acetylation of HSP90 and resultant targeting of JAK2 proteins for proteosomal degradation. This mechanism may contribute at least in part to the down-modulation of the JAK2V617F protein observed with the use of HDAC inhibitors.

Close modal

Clinical investigation of DNMT inhibitors in MF

The clinical investigation of epigenetic modulators in MF thus far has been limited to a few small, early phase trials involving DNMT or HDAC inhibitors. The clinical efficacy reported in the majority of published reports has been modest. In a phase 2 study of the DNMT inhibitor 5-azacytidine administered on a 7-day schedule in 34 patients with MF, the overall response rate according to International Working Group (IWG) criteria was 24%, including 1 partial response. The majority of responses were clinical improvement in spleen size.15  A similar smaller study was conducted by the Mayo Clinic with the same agent in 10 patients. In the latter study, 5-azacytidine was administered on a 5-day schedule. No responses were observed. Only 2 patients in that study received more than 3 cycles.16  The most common reasons for treatment discontinuation were disease progression and toxicity. Grade 3/4 adverse toxicities observed were mainly hematologic in nature and included neutropenia and thrombocytopenia.

The DNMT inhibitor 5-aza-2′-deoxyxytidine (decitabine) has also been investigated in MF. In a preliminary report of a multicenter phase 2 trial in which low-dose decitabine was administered subcutaneously on a 10-day schedule, responses including improvement in anemia and thrombocytopenia were observed in 7 of 19 evaluable patients. This was associated with a sustained decline in circulating CD34+ cells in responding patients. Toxicity was largely hematologic; there were few nonhematologic side effects.17  There have also been published case reports of clinical benefit associated with decitabine use in MF.18 

There is emerging evidence to suggest that DNMT inhibitors may be active in the treatment of Ph MPNs that have progressed to acute myeloid leukemia or myelodysplastic syndrome (MDS).19,20  This is a subgroup of patients with a very dismal outcome, typified by an extremely short survival with conventional treatment approaches including intensive chemotherapeutic regimens.21  The Groupe Francophone des Myelodysplasies (GFM) has reported on the use of azacitidine in 54 patients with Ph MPNs who had progressed to acute myeloid leukemia or MDS and reported a 52% overall response rate including 24% complete responses and 11% partial responses. Median response duration was 9 months, suggesting the need for additional alternative or consolidative approaches to produce further improvements in outcome.20 

Clinical investigation of HDAC inhibitors in MF

HDAC inhibitors have also been investigated in clinical trials in MF. In a phase 1 dose-escalation study of panobinostat in 18 patients with MF,22  3 of 5 evaluable patients who received more than 6 cycles of therapy had an improvement in spleen volume and/or anemia. Reversible thrombocytopenia was the dose-limiting toxicity. Interestingly, one of these patients was reported to have achieved a near complete remission with significant reduction in BM fibrosis and resolution of splenomegaly and leukoerythoblastosis, effects that are unusual for nontransplantation therapy in this disease. The recommended phase 2 dose in this trial was 25 mg three times weekly, which was significantly lower than the doses used in other early phase trials conducted with this agent. The higher doses used in the latter trials were23,24  associated with significant fatigue, diarrhea, and thrombocytopenia requiring dose reductions and leading to early treatment discontinuation.23,24  Other HDAC inhibitors that have been investigated in MF with demonstrable but modest activity include givinostat25  and pracinostat.26  Interestingly, the responses observed were independent of the JAK2 mutational status, suggesting that additional work is necessary to identify the predictors of clinical activity given the pleiotropic effects of these agents and the mutational complexity of MF.

Combination approaches involving epigenetic modulators in MF

The clinical trials conducted to date with DNMT and HDAC inhibitors give some overall signal of modest efficacy. The experience with this class of agents in other myeloid neoplasms such as MDS would suggest that repeated cycles are necessary for optimum clinical activity. Ongoing challenges include the fact that optimum doses and schedules in MF are not yet defined and that MF patients may be more susceptible to myelosuppression with these agents, which may be dose limiting. There are ongoing efforts to determine optimal combinations, potentially using lower doses that may synergize with other agents and confer improvement in patient outcomes.

There is preclinical evidence in vitro and in murine models to suggest several potentially rational combinations. These include combination of an HDAC inhibitor with a DNMT inhibitor to simultaneously effect 2 pathways of epigenetic silencing in MF.11  In addition, because the antitumor activity of HDAC inhibitors in MF is likely due at least in part to inhibition of HDAC6, with subsequent acetylation of HSP90 and targeting of oncogenic JAK proteins for degradation by the ubiquitin-proteosome system,14,24,27  rational combinations of HDAC inhibitors plus JAK inhibitors could also be explored. This approach is supported by preclinical studies demonstrating synergy in vitro in JAK2V617F mutant cell lines and in vivo in murine models when HDAC inhibitors are combined with JAK inhibitors.14,28  There are already clinical trials in progress (Table 2) investigating some of the aforementioned combinations.

Table 2.

Selected combination trials in clinical development in MF

Selected combination trials in clinical development in MF
Selected combination trials in clinical development in MF

Additional work is necessary to determine whether other components of the epigenetic regulation machinery, such as histone methyltransferases, histone demethylases, histone acetyltransferases, and sirtuins, are potential therapeutic targets in MF. Recent preclinical studies suggest that the effects of mutant proteins such as IDH1 and IDH2, which contribute to epigenetic dysregulation in myeloid neoplasia, can be abrogated by specific inhibitors of those proteins.29,30  The development of inhibitors targeted to mutated components of the epigenetic machinery has the potential to transform the epigenetic therapeutic landscape in myeloid neoplasms including MF.

Thalidomide analogs

Immunomodulatory drugs (ImiDs) are characterized by their ability to inhibit lipopolysaccharide-induced inflammatory cytokines such as TNFα, in addition to costimulation of T cells and augmentation of cytotoxic T- and NK-cell effector functions. The effects of these agents are pleiotropic and include antiangiogenic effects. More recently, epigenetic effects have also been demonstrated.31 

Response rates with thalidomide-based regimens when reassessed recently by IWG criteria were in the 22% range for anemia and 8% for splenomegaly, with a median duration of 8.5 months.32  Peripheral neuropathy, constipation, and myelosuppression were the most notable toxicities. Twenty-two percent of patients overall failed to complete 3 cycles of therapy and the thalidomide-prednisone regimen was the most tolerable of the 3 thalidomide-based regimens evaluated.32  In a multicenter phase 2 study of lenalidomide and prednisone in MF, response rate by IWG criteria was 23%; most of these were clinical improvement in anemia, with myelosuppression being the most significant toxicity encountered.33  Significant activity has been reported in individual cases of MF associated with the 5q cytogenetic aberration, which is a relatively rare subset of MF.34,35 

Pomalidomide is the newest thalidomide analog to be investigated in MF based on the premise that ImiDs have measurable clinical activity in MF. In a phase 2, multicenter, randomized, double-blind, adaptive design study of pomalidomide in MF,36  4 treatment arms were evaluated including pomalidomide 2 mg daily plus placebo, pomalidomide 2 mg daily plus prednisone, pomalidomide 0.5 mg daily plus prednisone, and prednisone plus placebo. Prednisone was administered on a tapering schedule for 3 cycles starting at 30 mg/d in the first cycle. The IWG anemia response rates in the 4 treatment arms were 23%, 16%, 36%, and 19%, respectively. Median duration of response was 7.5 months. Pomalidomide was well tolerated in that trial, with relatively minimal myelosuppression observed (in contrast to prior experience with lenalidomide), and there were no significant reports of peripheral neuropathy (in contrast to prior experience with thalidomide). Overall, the results suggested that the 0.5 mg dose was at least equivalent with regard to efficacy and potentially less toxic. A subsequent report of the long-term outcome of 94 patients enrolled in 2 consecutive pomalidomide trials conducted at a single institution revealed an IWG anemia response rate of 27%, with a median duration of response of 16 months. The absence of a JAK2V617F mutation in the context of circulating blasts ≥ 5% or palpable splenomegaly ≥ 10 cm was negatively correlated with the achievement of a response.37 

The results from these early phase trials provided a rationale for further investigation of pomalidomide in MF. Accrual has been completed for an ongoing placebo-controlled phase 3 trial of pomalidomide in transfusion-dependent MF (Table 1), and the results of this trial are eagerly awaited. Given the demonstrable activity of thalidomide analogs in improving anemia in MF, there are combination trials under way of ImiDs with other agents, including JAK inhibitors, in an effort to optimize the clinical activity of this class of drugs in MF (Table 2).

IFN

Multiple mechanisms have been proposed for IFN-α activity in myeloid neoplasia, including effects on immunomodulatory cells such as T cells and NK cells, induction of proapototic genes, suppression of proliferation of hematopoietic progenitor cells, and inhibition of angiogenesis.38  IFN-α at conventional doses in established MF is associated with significant myelosuppression and nonhematologic toxicities such as fatigue that have limited its use in MF. With the availability of more tolerable pegylated preparations and the demonstration that pegylated IFN can induce molecular responses in polycythemia vera, there has been a recent interest in investigating IFN in MF, particularly in MF patients in whom there is still a relative preservation of hematopoietic function.39,40  The clinical experience with IFN has recently been reviewed in detail.38  Larger prospective randomized trials (Table 1) will be required to validate the activity of this agent in early PMF, particularly with regard to the controversy surrounding the potential for reversal of BM histomorphologic changes with this agent.

Heat shock protein 90 (HSP90) serves as a molecular chaperone, resulting in the stabilization of several oncoproteins, so this molecule is often regarded as a facilitator of oncogenic addiction in various malignancies.41  HSP90 inhibition results in the targeting of oncogenic client proteins for proteosomal degradation, so HSP90 has emerged as a rational therapeutic target in various neoplasms. Both wild-type JAK2 and JAK2V617F have been demonstrated to be client proteins of HSP90.42  In addition, in vitro treatment of MPN cell lines and primary patient samples with HSP90 inhibitors resulted in degradation of JAK2 protein, inhibition of JAK/STAT signaling, and inhibition of cell growth.42,43 

In JAK2 mutant murine models of PV and ET, in addition to the above effects, HSP90 inhibition led to normalization of peripheral blood counts, decline of the JAK2V617F allele burden, and improved survival in mice at a dose that did not affect JAK2 in normal tissues or cause significant toxicity.42  Interestingly, heterodimeric JAK/STAT activation (heterodimerization of JAK2 with JAK1 or TYK2) has been implicated as a mechanism of persistent disease in the face of JAK2 inhibitor therapy and cell lines that harbored this mechanism of activated JAK2 persistence retained their sensitivity to HSP90 inhibitors.44  In addition, cells engineered to express JAK2 kinase domain mutations that conferred resistance to JAK-ATP mimetic inhibitors also retained their sensitivity to HSP90 inhibitors.45  These findings provide a compelling rationale for the clinical investigation of HSP90 inhibitors in MF.

There is currently an ongoing single-agent trial of the HSP90 inhibitor AUY922 in patients with Ph− MPNs, including MF (Table 1). Combination studies of HSP90 plus JAK inhibitors are also reasonable based on potential additive effects demonstrated preclinically between these agents.42 

Allogeneic stem cell transplantation is the only therapeutic modality to date that has been demonstrated conclusively to lead to a reversal of fibrosis in MF. The pathogenetic mechanisms underlying the development of fibrosis in MF are poorly understood. Although MF has been established to be a clonal disorder of pluripotent stem cell origin, the fibroblasts have been shown to be polyclonal and are functionally similar to normal fibroblasts. The BM findings in MF include increases in the numbers of stromal cells and in levels of extracellular matrix proteins, angiogenesis, and osteosclerosis.

Monoclonal antibodies

A current pathogenetic hypothesis with regard to the genesis of fibrosis in MF is that clonal megakaryocytes secrete fibrogenic and angiogenic cytokines including TGFβ and MMP-9.46,47  A common theme emerging from several murine models of MF is that there is a significant association between increased numbers of megakaryocytes and the development of an extracellular matrix typical of MF, and these findings lend credence to the hypothesis that megakaryocytes play a central role in fibrogenesis.47  The availability of novel agents targeting specific pathways and cytokines implicated in fibrogenesis has led to a resurgence in interest with regard to the clinical investigation of antifibrosis agents in MF.

For example, a monoclonal inhibitor of TGFβ, fresolimumab (GC-1008), was recently investigated in a phase 1 trial in MF48  and was found to be associated with a significant decline in TGFβ1 levels in both evaluable patients enrolled in that trial. The trial was terminated early due to drug supply issues after enrollment of just 3 patients. The potential merit of targeting TGFβ in MF is supported by a recent study identifying abnormal genetic signatures in TGFβ1 signaling in the spleen and BM from the GATA-1low mouse, a murine model of MF.49  Inhibition of TGFβ1 signaling in this mouse model normalized this aberrant gene expression signature, restored hematopoiesis and megakaryocyte development, and reduced fibrosis, neovascularization, and osteogenesis in the BM.49 

A different monoclonal antibody, simtuzumab (GS-6624), is being studied in a phase 2 clinical trial in MF. Simtuzumab is a monoclonal antibody against lysyl oxidase (LOX)-like protein 2. LOX is a copper-dependent enzyme that initiates the covalent cross-linking of collagen or elastin and was shown to be abundant in the GATA-1low mouse, which is characterized by significant MF and high levels of low-ploidy megakaryocytes. Inhibition of LOX enzymatic activity led to a significant improvement of the fibrotic phenotype, leading to the speculation that LOX is a potential therapeutic target in diseases such as MF.47,50 

Aurora kinase inhibitors

Another potential target of the megakaryocyte-fibrosis axis are aurora kinases. In acute megakaryocytic leukemia, a disease with a dismal prognosis characterized by expansion of immature megakaryocytes and profound BM fibrosis, aurora kinase A was identified as a mediator of polyploidization of malignant megakaryocytes. Aurora kinase A inhibition in acute megakaryocytic leukemia led to apoptosis of malignant megakaryocytes and induced polyploidization and expression of mature megakaryocyte markers.51  Given the prominent role of clonal megakaryocytes in MF, these findings have led to the speculation that aurora kinase A could be a valid therapeutic target in MF, and there are ongoing preclinical studies to validate these assertions.

There several signaling intermediates downstream of the JAK/STAT signaling pathway (Figure 3) that constitute rational therapeutic targets in MF. The PI3K/AKT/mammalian target of rapamycin (mTOR) pathway has been shown to be dysregulated in MPNs and AKT activation has been found to be critical for JAK2V617F-mediated cellular transformation. AKT and mTOR inhibitors have been associated with growth inhibition of primary MPN cells and cell lines in vitro. These findings have led to the clinical investigation of mTOR inhibitors in MF.52  In a phase 1/2 study of everolimus in MF, 20% of patients achieved a rapid/sustained reduction in splenomegaly of > 50% and more than two-thirds of patients enrolled experienced complete resolution of constitutional symptoms and improvement in pruritus. A minority of patients (15%-25%) experienced improvement in anemia and thrombocytopenia. Although known targets of mTOR such as phosphor-p70S6K were identified as potential biomarkers of response to the agent, there was no significant effect on MF-related biomarkers such as JAK2V617F allelic burden, circulating CD34+ cells, or cytokine levels. The drug was well tolerated, with grade 1-2 stomatitis being the most common toxicity.

Figure 3.

Novel agents targeting pathways downstream of the JAK/STAT signaling pathway in MF. Activated JAK2 signals through and activates downstream signaling intermediates such as STAT5, RAS/MAPK, and PI3K/AKT/mTOR pathways, leading to effects on proliferation and survival of MPN cells. JAK proteins can be down-modulated by the use of HSP90 inhibitors or HDAC inhibitors, which lead to targeting of both wild-type and mutant proteins for degradation by the proteosomal system. PI3K/AKT inhibitors, mTOR inhibitors, MEK inhibitors, and STAT inhibitors can inhibit the respective signaling intermediates downstream of JAK/STAT pathway. DNMT inhibitors can potentially reverse epigenetic silencing of various genes including the SOCS genes, which are negative regulators of the JAK/STAT signaling pathway.

Figure 3.

Novel agents targeting pathways downstream of the JAK/STAT signaling pathway in MF. Activated JAK2 signals through and activates downstream signaling intermediates such as STAT5, RAS/MAPK, and PI3K/AKT/mTOR pathways, leading to effects on proliferation and survival of MPN cells. JAK proteins can be down-modulated by the use of HSP90 inhibitors or HDAC inhibitors, which lead to targeting of both wild-type and mutant proteins for degradation by the proteosomal system. PI3K/AKT inhibitors, mTOR inhibitors, MEK inhibitors, and STAT inhibitors can inhibit the respective signaling intermediates downstream of JAK/STAT pathway. DNMT inhibitors can potentially reverse epigenetic silencing of various genes including the SOCS genes, which are negative regulators of the JAK/STAT signaling pathway.

Close modal

Potent inhibitors of the PI3K/AKT pathway are entering into clinical trials in MF. Combinations of JAK inhibitors and PI3K inhibitors may also be reasonable (Table 2) based on emerging preclinical evidence that suggests synergy.

Given the variability in outcome associated with MF, a comprehensive assessment including risk stratification according to contemporary prognostic scoring models such as the Dynamic International Prognostic Scoring System (DIPSS)53,54 or DIPSS plus55  is essential to help guide treatment recommendations. I use a watch and wait approach in asymptomatic lower (low- and intermediate-1) risk patients, whereas those with higher (intermediate-2 and high-) risk disease require intervention (Figure 4). Given the emerging data for improved tolerance with reduced intensity conditioning transplantation regimens, early referral for allogeneic stem cell transplantation should be considered in transplantation-eligible patients.56  I recommend JAK inhibitor therapy for symptomatic patients with MF, particularly in the absence of significant cytopenias. In the presence of significant cytopenias or for patients with prior JAK inhibitor exposure, I enroll patients in a clinical trial whenever possible. For MF-accelerated or blast phase disease, a particularly poor prognostic subset of patients,21  immediate referral for allogeneic stem cell transplantation in transplantation-eligible patients should be strongly considered. In these instances, I recommend hypomethylating agent therapy17,19,20  whenever feasible or cytarabine-based induction approaches for cytoreduction and/or conversion to a second chronic phase of the disease before proceeding with transplantation.

Figure 4.

How I treat MF in the JAK inhibitor era. The DIPSS can be used for risk stratification at any time point during the course of MF. Prognostic variables are each assigned a score of 1 point, except for anemia, which is assigned a score of 2 points. These variables include: age > 65 years, anemia (defined as hemoglobin < 10 g/dL), WBC count > 25 × 109/L, circulating blasts ≥ 1%, and constitutional systems. The recommended treatment algorithm according to risk stratum categories (low, intermediate 1 [Int-1]; intermediate 2 [Int-2], and high) is summarized in the figure. Early referral for allogeneic stem cell transplantation and/or enrollment in clinical trials is recommended for symptomatic or higher risk patients with significant cytopenias (hemoglobin < 10 g/dL, platelet count < 100 K/μL) and/or prior JAK inhibitor exposure. In the absence of an appropriate clinical trial, ImiD agents, androgenic steroids, or erythropoietin-stimulating agents can be considered, particularly for anemic patients.

Figure 4.

How I treat MF in the JAK inhibitor era. The DIPSS can be used for risk stratification at any time point during the course of MF. Prognostic variables are each assigned a score of 1 point, except for anemia, which is assigned a score of 2 points. These variables include: age > 65 years, anemia (defined as hemoglobin < 10 g/dL), WBC count > 25 × 109/L, circulating blasts ≥ 1%, and constitutional systems. The recommended treatment algorithm according to risk stratum categories (low, intermediate 1 [Int-1]; intermediate 2 [Int-2], and high) is summarized in the figure. Early referral for allogeneic stem cell transplantation and/or enrollment in clinical trials is recommended for symptomatic or higher risk patients with significant cytopenias (hemoglobin < 10 g/dL, platelet count < 100 K/μL) and/or prior JAK inhibitor exposure. In the absence of an appropriate clinical trial, ImiD agents, androgenic steroids, or erythropoietin-stimulating agents can be considered, particularly for anemic patients.

Close modal

It has been many decades since Damashek's original description of the MPNs, and MF has remained a largely incurable disease. The discovery of the JAK2V617F mutation and the development of JAK inhibitors have been a significant step forward, and have fueled the resurgence of interest in this group of diseases. Recent biologic insights underscore the molecular complexity of this group of diseases and it will be critical to decipher which genetic and epigenetic aberrations are the key drivers. This knowledge, along with an increasing plethora of new agents that target pathways of potential pathogenetic importance in MF, is likely to bring us closer to an agent or agents that will significantly change the natural history of this disease.

Conflict-of-interest disclosure: The author is on the board of directors or an advisory committee for Suneisis Pharmaceuticals, Spectrum Pharmaceuticals, Sanofi-Aventis, and Incyte and has received research funding from Topotarget and Eisai. Off-label drug use: None disclosed.

Olatoyosi Odenike, Department of Medicine and Comprehensive Cancer Center, The University of Chicago, 5841 South Maryland Ave, Chicago, IL 60637; Phone: 773-702-3354; Fax: 773-834-0188; e-mail: todenike@medicine.bsd.uchicago.edu.

1
Harrison
 
C
Kiladjian
 
JJ
Al-Ali
 
HK
, et al. 
JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis
N Engl J Med
2012
, vol. 
366
 
9
(pg. 
787
-
798
)
2
Verstovsek
 
S
Mesa
 
RA
Gotlib
 
J
, et al. 
A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis
N Engl J Med
2012
, vol. 
366
 
9
(pg. 
799
-
807
)
3
Mascarenhas
 
J
Hoffman
 
R
A comprehensive review and analysis of the effect of ruxolitinib therapy on the survival of patients with myelofibrosis
Blood
2013
, vol. 
121
 
24
(pg. 
4832
-
4837
)
4
Chen
 
J
Odenike
 
O
Rowley
 
JD
Leukaemogenesis: more than mutant genes
Nat Rev Cancer
2010
, vol. 
10
 
1
(pg. 
23
-
36
)
5
Teofili
 
L
Martini
 
M
Cenci
 
T
, et al. 
Epigenetic alteration of SOCS family members is a possible pathogenetic mechanism in JAK2 wild-type myeloproliferative diseases
Int J Cancer
2008
, vol. 
123
 
7
(pg. 
1586
-
1592
)
6
Bogani
 
C
Ponziani
 
V
Guglielmelli
 
P
, et al. 
Hypermethylation of CXCR4 promoter in CD34+ cells from patients with primary myelofibrosis
Stem Cells
2008
, vol. 
26
 
8
(pg. 
1920
-
1930
)
7
Vannucchi
 
AM
Lasho
 
TL
Guglielmelli
 
P
, et al. 
Mutations and prognosis in primary myelofibrosis
Leukemia
 
Published online ahead of print April 26, 2013. doi:10.1038/leu.2013.119
8
Dawson
 
MA
Bannister
 
AJ
Gottgens
 
B
, et al. 
JAK2 phosphorylates histone H3Y41 and excludes HP1alpha from chromatin
Nature
2009
, vol. 
461
 
7265
(pg. 
819
-
822
)
9
Liu
 
F
Zhao
 
X
Perna
 
F
, et al. 
JAK2V617F-mediated phosphorylation of PRMT5 downregulates its methyltransferase activity and promotes myeloproliferation
Cancer Cell
2011
, vol. 
19
 
2
(pg. 
283
-
294
)
10
Nischal
 
S
Bhattacharyya
 
S
Christopeit
 
M
, et al. 
Methylome profiling reveals distinct alterations in phenotypic and mutational subgroups of myeloproliferative neoplasms
Cancer Res
2013
, vol. 
73
 
3
(pg. 
1076
-
1085
)
11
Wang
 
X
Zhang
 
W
Ishii
 
T
, et al. 
Correction of the abnormal trafficking of primary myelofibrosis CD34+ cells by treatment with chromatin-modifying agents
Cancer Res
2009
, vol. 
69
 
19
(pg. 
7612
-
7618
)
12
Guerini
 
V
Barbui
 
V
Spinelli
 
O
, et al. 
The histone deacetylase inhibitor ITF2357 selectively targets cells bearing mutated JAK2(V617F)
Leukemia
2008
, vol. 
22
 
4
(pg. 
740
-
747
)
13
Shang
 
L
Tomasi
 
TB
The heat shock protein 90-CDC37 chaperone complex is required for signaling by types I and II interferons
J Biol Chem
2006
, vol. 
281
 
4
(pg. 
1876
-
1884
)
14
Wang
 
Y
Fiskus
 
W
Chong
 
DG
, et al. 
Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells
Blood
2009
, vol. 
114
 
24
(pg. 
5024
-
5033
)
15
Quintas-Cardama
 
A
Tong
 
W
Kantarjian
 
H
, et al. 
A phase II study of 5-azacitidine for patients with primary and post-essential thrombocythemia/polycythemia vera myelofibrosis
Leukemia
2008
, vol. 
22
 
5
(pg. 
965
-
970
)
16
Mesa
 
RA
Verstovsek
 
S
Rivera
 
C
, et al. 
Bortezomib therapy in myelofibrosis: a phase II clinical trial
Leukemia
2008
, vol. 
22
 
8
(pg. 
1636
-
1638
)
17
Odenike
 
OM
Godwin
 
JE
Van Besien
 
K
, et al. 
Phase II trial of low dose, subcutaneous decitabine in myelofibrosis [abstract]
Blood (ASH Annual Meeting Abstracts)
2008
, vol. 
112
 
11
pg. 
2809
 
18
Liu
 
Y
Tabarroki
 
A
Billings
 
S
, et al. 
Successful use of very low dose subcutaneous decitabine to treat high-risk myelofibrosis with Sweet's syndrome that was refractory to 5-azacitidine
Leuk Lymphoma
 
Published online ahead of print June 24, 2013. doi:10.3109/10428194.2013.802315
19
Mascarenhas
 
J
Navada
 
S
Malone
 
A
Rodriguez
 
A
Najfeld
 
V
Hoffman
 
R
Therapeutic options for patients with myelofibrosis in blast phase
Leuk Res
2010
, vol. 
34
 
9
(pg. 
1246
-
1249
)
20
Thepot
 
S
Itzykson
 
R
Seegers
 
V
, et al. 
Treatment of progression of Philadelphia-negative myeloproliferative neoplasms to myelodysplastic syndrome or acute myeloid leukemia by azacitidine: a report on 54 cases on the behalf of the Groupe Francophone des Myelodysplasies (GFM)
Blood
2010
, vol. 
116
 
19
(pg. 
3735
-
3742
)
21
Mesa
 
RA
Li
 
CY
Ketterling
 
RP
Schroeder
 
GS
Knudson
 
RA
Tefferi
 
A
Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases
Blood
2005
, vol. 
105
 
3
(pg. 
973
-
977
)
22
Mascarenhas
 
J
Lu
 
M
Li
 
T
, et al. 
A phase I study of panobinostat (LBH589) in patients with primary myelofibrosis (PMF) and post-polycythaemia vera/essential thrombocythaemia myelofibrosis (post-PV/ET MF)
Br J Haematol
2013
, vol. 
161
 
1
(pg. 
68
-
75
)
23
Deangelo
 
DJ
Spencer
 
A
Bhalla
 
KN
, et al. 
Phase Ia/II, two-arm, open-label, dose-escalation study of oral panobinostat administered via two dosing schedules in patients with advanced hematologic malignancies
Leukemia
2013
, vol. 
27
 
8
(pg. 
1628
-
1636
)
24
Deangelo
 
DJ
Mesa
 
RA
Fiskus
 
W
, et al. 
Phase II trial of panobinostat, an oral pan-deacetylase inhibitor in patients with primary myelofibrosis, post-essential thrombocythaemia, and post-polycythaemia vera myelofibrosis
Br J Haematol
2013
, vol. 
162
 
3
(pg. 
326
-
335
)
25
Rambaldi
 
A
Dellacasa
 
CM
Finazzi
 
G
, et al. 
A pilot study of the Histone-Deacetylase inhibitor Givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms
Br J Haematol
2010
, vol. 
150
 
4
(pg. 
446
-
455
)
26
Quintas-Cardama
 
A
Kantarjian
 
H
Estrov
 
Z
Borthakur
 
G
Cortes
 
J
Verstovsek
 
S
Therapy with the histone deacetylase inhibitor pracinostat for patients with myelofibrosis
Leuk Res
2012
, vol. 
36
 
9
(pg. 
1124
-
1127
)
27
Bali
 
P
Pranpat
 
M
Bradner
 
J
, et al. 
Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors
J Biol Chem
2005
, vol. 
280
 
29
(pg. 
26729
-
26734
)
28
Baffert
 
F
Evrot
 
E
Ebel
 
N
, et al. 
Improved efficacy upon combined JAK1/2 and pan-deacetylase inhibition using ruxolitinib (INC424) and panobinostat (LBH589) in preclinical mouse models of JAK2V617F-driven disease [abstract]
Blood (ASH Annual Meeting Abstracts)
2011
, vol. 
118
 
21
pg. 
798
 
29
Losman
 
JA
Looper
 
RE
Koivunen
 
P
, et al. 
(R)-2-hydroxyglutarate is sufficient to promote leukemogenesis and its effects are reversible
Science
2013
, vol. 
339
 
6127
(pg. 
1621
-
1625
)
30
Wang
 
F
Travins
 
J
DeLaBarre
 
B
, et al. 
Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation
Science
2013
, vol. 
340
 
6132
(pg. 
622
-
626
)
31
Shortt
 
J
Hsu
 
AK
Johnstone
 
RW
Thalidomide-analogue biology: immunological, molecular and epigenetic targets in cancer therapy
Oncogene
 
Published online ahead of print June 14, 2013. doi:10.1038/onc.2012.599
32
Thapaliya
 
P
Tefferi
 
A
Pardanani
 
A
, et al. 
International working group for myelofibrosis research and treatment response assessment and long-term follow-up of 50 myelofibrosis patients treated with thalidomide-prednisone based regimens
Am J Hematol
2011
, vol. 
86
 
1
(pg. 
96
-
98
)
33
Mesa
 
RA
Yao
 
X
Cripe
 
LD
, et al. 
Lenalidomide and prednisone for myelofibrosis: Eastern Cooperative Oncology Group (ECOG) phase 2 trial E4903
Blood
2010
, vol. 
116
 
22
(pg. 
4436
-
4438
)
34
Takahashi
 
K
Cortes
 
J
Pierce
 
S
Abruzzo
 
L
Kantarjian
 
H
Verstovsek
 
S
Chromosome 5q deletion is extremely rare in patients with myelofibrosis
Leuk Res
2013
, vol. 
37
 
5
(pg. 
552
-
555
)
35
Tefferi
 
A
Lasho
 
TL
Mesa
 
RA
Pardanani
 
A
Ketterling
 
RP
Hanson
 
CA
Lenalidomide therapy in del(5)(q31)-associated myelofibrosis: cytogenetic and JAK2V617F molecular remissions
Leukemia
2007
, vol. 
21
 
8
(pg. 
1827
-
1828
)
36
Tefferi
 
A
Verstovsek
 
S
Barosi
 
G
, et al. 
Pomalidomide is active in the treatment of anemia associated with myelofibrosis
J Clin Oncol
2009
, vol. 
27
 
27
(pg. 
4563
-
4569
)
37
Begna
 
KH
Pardanani
 
A
Mesa
 
R
, et al. 
Long-term outcome of pomalidomide therapy in myelofibrosis
Am J Hematol
2012
, vol. 
87
 
1
(pg. 
66
-
68
)
38
Kiladjian
 
JJ
Mesa
 
RA
Hoffman
 
R
The renaissance of interferon therapy for the treatment of myeloid malignancies
Blood
2011
, vol. 
117
 
18
(pg. 
4706
-
4715
)
39
Ianotto
 
JC
Kiladjian
 
JJ
Demory
 
JL
, et al. 
PEG-IFN-alpha-2a therapy in patients with myelofibrosis: a study of the French Groupe d'Etudes des Myelofibroses (GEM) and France Intergroupe des syndromes Myeloproliferatifs (FIM)
Br J Haematol
2009
, vol. 
146
 
2
(pg. 
223
-
225
)
40
Silver
 
RT
Vandris
 
K
Goldman
 
JJ
Recombinant interferon-alpha may retard progression of early primary myelofibrosis: a preliminary report
Blood
2011
, vol. 
117
 
24
(pg. 
6669
-
6672
)
41
Trepel
 
J
Mollapour
 
M
Giaccone
 
G
Neckers
 
L
Targeting the dynamic HSP90 complex in cancer
Nat Rev Cancer
2010
, vol. 
10
 
8
(pg. 
537
-
549
)
42
Marubayashi
 
S
Koppikar
 
P
Taldone
 
T
, et al. 
HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans
J Clin Invest
2010
, vol. 
120
 
10
(pg. 
3578
-
3593
)
43
Bareng
 
J
Jilani
 
I
Gorre
 
M
, et al. 
A potential role for HSP90 inhibitors in the treatment of JAK2 mutant-positive diseases as demonstrated using quantitative flow cytometry
Leuk Lymphoma
2007
, vol. 
48
 
11
(pg. 
2189
-
2195
)
44
Koppikar
 
P
Bhagwat
 
N
Kilpivaara
 
O
, et al. 
Heterodimeric JAK-STAT activation as a mechanism of persistence to JAK2 inhibitor therapy
Nature
2012
, vol. 
489
 
7414
(pg. 
155
-
159
)
45
Weigert
 
O
Lane
 
AA
Bird
 
L
, et al. 
Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition
J Exp Med
2012
, vol. 
209
 
2
(pg. 
259
-
273
)
46
Ciurea
 
SO
Merchant
 
D
Mahmud
 
N
, et al. 
Pivotal contributions of megakaryocytes to the biology of idiopathic myelofibrosis
Blood
2007
, vol. 
110
 
3
(pg. 
986
-
993
)
47
Papadantonakis
 
N
Matsuura
 
S
Ravid
 
K
Megakaryocyte pathology and bone marrow fibrosis: the lysyl oxidase connection
Blood
2012
, vol. 
120
 
9
(pg. 
1774
-
1781
)
48
Mascarenhas
 
J
Li
 
T
Sandy
 
L
, et al. 
Anti-transforming growth factor beta (TGF-beta) therapy in patients with myelofibrosis
Leuk Lymphoma
 
Published online ahead of print June 24, 2013. doi:10.3109/10428194.2013.802315
49
Zingariello
 
M
Martelli
 
F
Ciaffoni
 
F
, et al. 
Characterization of the TGF-beta1 signaling abnormalities in the Gata1low mouse model of myelofibrosis
Blood
2013
, vol. 
121
 
17
(pg. 
3345
-
3363
)
50
Eliades
 
A
Papadantonakis
 
N
Bhupatiraju
 
A
, et al. 
Control of megakaryocyte expansion and bone marrow fibrosis by lysyl oxidase
J Biol Chem
2011
, vol. 
286
 
31
(pg. 
27630
-
27638
)
51
Wen
 
Q
Goldenson
 
B
Silver
 
SJ
, et al. 
Identification of regulators of polyploidization presents therapeutic targets for treatment of AMKL
Cell
2012
, vol. 
150
 
3
(pg. 
575
-
589
)
52
Guglielmelli
 
P
Barosi
 
G
Rambaldi
 
A
, et al. 
Safety and efficacy of everolimus, a mTOR inhibitor, as single agent in a phase 1/2 study in patients with myelofibrosis
Blood
2011
, vol. 
118
 
8
(pg. 
2069
-
2076
)
53
Passamonti
 
F
Cervantes
 
F
Vannucchi
 
AM
, et al. 
A dynamic prognostic model to predict survival in primary myelofibrosis: a study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment)
Blood
2010
, vol. 
115
 
9
(pg. 
1703
-
1708
)
54
Passamonti
 
F
Cervantes
 
F
Vannucchi
 
AM
, et al. 
Dynamic International Prognostic Scoring System (DIPSS) predicts progression to acute myeloid leukemia in primary myelofibrosis
Blood
2010
, vol. 
116
 
15
(pg. 
2857
-
2858
)
55
Gangat
 
N
Caramazza
 
D
Vaidya
 
R
, et al. 
DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status
J Clin Oncol
2011
, vol. 
29
 
4
(pg. 
392
-
397
)
56
Gupta
 
V
Hari
 
P
Hoffman
 
R
Allogeneic hematopoietic cell transplantation for myelofibrosis in the era of JAK inhibitors
Blood
2012
, vol. 
120
 
7
(pg. 
1367
-
1379
)